ArticlePDF Available

Agonistic Properties of Cannabidiol at 5-HT1a Receptors

Authors:
  • CReDO Science
  • University of Nevada Reno School of Medicine

Abstract and Figures

Cannabidiol (CBD) is a major, biologically active, but psycho-inactive component of cannabis. In this cell culture-based report, CBD is shown to displace the agonist, [3H]8-OH-DPAT from the cloned human 5-HT1a receptor in a concentration-dependent manner. In contrast, the major psychoactive component of cannabis, tetrahydrocannabinol (THC) does not displace agonist from the receptor in the same micromolar concentration range. In signal transduction studies, CBD acts as an agonist at the human 5-HT1a receptor as demonstrated in two related approaches. First, CBD increases [35S]GTPgammaS binding in this G protein coupled receptor system, as does the known agonist serotonin. Second, in this GPCR system, that is negatively coupled to cAMP production, both CBD and 5-HT decrease cAMP concentration at similar apparent levels of receptor occupancy, based upon displacement data. Preliminary comparative data is also presented from the cloned rat 5-HT2a receptor suggesting that CBD is active, but less so, relative to the human 5-HT1a receptor, in binding analyses. Overall, these studies demonstrate that CBD is a modest affinity agonist at the human 5-HT1a receptor. Additional work is required to compare CBD's potential at other serotonin receptors and in other species. Finally, the results indicate that cannabidiol may have interesting and useful potential beyond the realm of cannabinoid receptors.
Content may be subject to copyright.
Agonistic Properties of Cannabidiol at 5-HT1a Receptors
Ethan B. Russo,
1
Andrea Burnett,
1
Brian Hall,
1
and Keith K. Parker
1,2
(Accepted June 27, 2005)
Cannabidiol (CBD) is a major, biologically active, but psycho-inactive component of
cannabis. In this cell culture-based report, CBD is shown to displace the agonist, [3H]8-OH-
DPAT from the cloned human 5-HT1a receptor in a concentration-dependent manner. In
contrast, the major psychoactive component of cannabis, tetrahydrocannabinol (THC) does
not displace agonist from the receptor in the same micromolar concentration range. In signal
transduction studies, CBD acts as an agonist at the human 5-HT1a receptor as demonstrated
in two related approaches. First, CBD increases [35S]GTPcS binding in this G protein coupled
receptor system, as does the known agonist serotonin. Second, in this GPCR system, that is
negatively coupled to cAMP production, both CBD and 5-HT decrease cAMP concentration
at similar apparent levels of receptor occupancy, based upon displacement data. Preliminary
comparative data is also presented from the cloned rat 5-HT2a receptor suggesting that CBD
is active, but less so, relative to the human 5-HT1a receptor, in binding analyses. Overall, these
studies demonstrate that CBD is a modest affinity agonist at the human 5-HT1a receptor.
Additional work is required to compare CBD’s potential at other serotonin receptors and in
other species. Finally, the results indicate that cannabidiol may have interesting and useful
potential beyond the realm of cannabinoid receptors.
KEY WORDS: Cannabis; cannabidiol; cAMP; G Proteins; marijuana; serotonin; THC.
INTRODUCTION
Although cannabis and its extracts have been
extensively studied, knowledge of the biochemical
mechanisms of one of its major components, canna-
bidiol (CBD), has not been thoroughly explored (1,2).
This lack of knowledge of CBD’s biochemical phar-
macology is noteworthy in the context of its known
potential in human therapy: for example, it has been
demonstrated to have anxiolytic (3), anti-seizure (4),
anti-psychotic (3), and neuroprotective properties
(5,6). While previously thought to be sedating, recent
clinical research has confirmed that CBD is activat-
ing, and that it counters sedative effects of THC (7).
The major psychoactive component of cannabis,
tetrahydrocannabinol (THC), has received extensive
research attention into its biochemical pharmacology.
Both THC and CBD have been pharmacologically
investigated at cannabinoid receptors (CBR), which
are highly conserved across animal taxa, with the
major exception of insects (8–10). THC is at least 10
times more potent in binding to CB1 receptors than
CB2 receptors. At CB1R, there is evidence to suggest
that CBD is an antagonist or inverse agonist, al-
though substantial debate still exits about its intrinsic
activity (10,11). CBD has received little attention in
other neurotransmitter systems. Noteworthy in this
regard is serotonin (5-hydroxytryptamine; 5-HT),
which is known to be involved in many of the same
processes important to cannabis’s actions (12,13) such
as relief of anxiety, pain, the complex processes of
1
Skaggs School of Pharmacy, The University of Montana,
Missoula, MT 59812-1552, USA.
2
Address reprint requests to: Keith K. Parker, Skaggs School of
Pharmacy, The University of Montana, Missoula, MT 59812-
1552, USA. Tel.: +406-243-4235; Fax: +406-243-5228; E-mail:
keith.parker@umontana.edu
Neurochemical Research, Vol. 30, No. 8, August 2005 (Ó 2005), pp. 1037–1043
DOI: 10.1007/s11064-005-6978-1
1037
0364-3190/05/0800–1037/0 Ó 2005 Springer Science+Business Media, Inc.
headache (14,15), and thermoregulation. The few
studies done with CBD in serotonergic systems
suggest that it inhibits 5-HT re-uptake, and overall
reduces 5-HT neurotransmission (2,16). There is also
some experimental evidence to support CBD’s activity
in other neurotransmitter systems such as dopamine,
GABA, and the endogenous opioid system (2).
Most of 5-HT’s broad actions are thought to be
regulated at a series of 5-HT receptors (5-HTR), the
majority of which (17) are members of the diverse
super family of G-protein coupled (GPC), seven-
transmembrane receptors (7TMR). The 5-HT1aR
(17) has been cloned and studied in numerous in vivo
and cell culture systems and in various species. It has
been cloned in both human (H) and rat (18–20),
amongst other organisms, and has been further
analyzed in other species, including rabbit (21), where
it has not been cloned. In this literature, extending
over two decades, 5-HT1aR has been ever more
implicated in a variety of physiological and
pathological processes including anxiety, mood,
depression, panic, obsessive-compulsive disorders,
headache, immune regulation, and cardiovascular
regulation to name a few (2,6,17,18). Additionally, the
5-HT2aR could have relevance to the pharmacology
of cannabis as it has been associated with phenomena
like mood, headache, and hallucination (22). There is
precedence for the action of cannabinoids such as
oleamide at serotonin receptors (23–26).
Over the last decade our laboratory has con-
ducted a series of studies with 5-HT1aR (27), and to a
lesser extent with 5-HT2aR (21). Because of these
interests and our hypothesis that CBD may have
important actions relevant to the pharmacology of
cannabis but outside the realm of CBR, we report
here studies with H5-HT1aR a nd a limited compar-
ison to the rat 5-HT2aR (28). For both H5-HT1aR
and rat 5-HT2aR we also report comparisons be-
tween CBD and THC. In cell culture experiments
with cloned human 5-HT1aR and rat 5-HT2aR,
CBD has a greater affinity than THC for both
receptors. CBD binds with higher affinity at 5-
HT1aR than at 5-HT2aR. In the case of H5-HT1aR,
CBD appears to act as an agonist. A preliminary
report of these investigations has appeared (29).
EXPERIMENTAL PROCEDURE
Cell Culture. Chinese Hamster Ovary (CHO) cells expressing
the H5-HT1aR (19) were cultured in Ham’s F-12 medium fortified
with 10 % fetal calf serum and 200 ug/ml geneticin. Cultures were
maintained at 37°C in a humidified atmosphere of 5% CO2. Cells
were sub-cultured or assayed upon confluency (5–8 days). Cloned
H5-HT1aR was kindly provided by Dr. John Raymond (Medical
U. of South Carolina). NIH 3T3 cells expressing the rat 5-HT2aR
(28) were cultured under similar conditions in DMEM fortified
with 10% calf serum and 200 lg/ml geneticin. These transfected
cells were generously provided by Dr. David Julius (UCSF). Both
cell lines have been tested for mycoplasma with a PCR kit (ATCC),
and are free of contamination.
Receptor Preparation. Cells were harvested by trypsinization
and centrifuged at low speed in ice-cold medium. The pellet was re-
suspended in ice-cold Earle’s Balanced Salt Solution followed by
centrifugation. Cells were re-suspended in 10 ml of ice-cold binding
buffer (50 mM Tris, 4 mM CaCl2, 10 lM pargyline, pH 7.4),
homogenized with Teflon-glass, and centrifuged for 450,000 g-min.
at 4°C. To produce a crude membrane preparation, the pellet was
re-suspended in 30 ml of ice-cold binding buffer, and homogenized,
first with Teflon-glass and then with a Polytron (setting 4) for 5 s.
The receptor preparation was stored on ice and assayed within the
next 1.5 h.
Assay of Receptor Activity. Binding of the agonist [3H]8-OH-
DPAT ([3H]8-hydroxy-2-(di-n-propylamino)tetralin) to H5-
HT1aR followed well-characterized in vitro protocols (20,27,30).
Radioligands were purchased from New England Nuclear (NEN),
Boston, MA. 1 ml reaction mixtures, in triplicate, were incubated
for 30 min. in a 30°C shaker bath. Composition of the 1 ml reac-
tion mixture was: 700 ll of receptor preparation; 100 ll of either
binding buffer (for total binding) or 10 lM 5-HT (final concen-
tration for non-specific binding), 100 ll of the tritiated agent (final
concentration of 0.5 nM [3H] 8-OH-DPAT), and 100 ll of diluted
CBD or binding buffer in the case of controls.
Reactions were stopped by addition of 4 ml of ice-cold
50 mM Tris buffer, pH 7.4, and subsequent vacuum filtration on
glass fiber filters (Whatman GF/B). Filters were rinsed twice in
5 ml of ice-cold Tris buffer, dried, and counted in 5 ml of Ecoscint
(National Diagnostics) liquid scintillation fluid in a Beckman LS
6500 instrument. Homogenates were assayed for protein to main-
tain a nominal value of 50 lg protein per filter over weekly assays
(31). Total and non-specific binding tubes were run in triplicate.
Assays of the rat 5-HT2aR (28) were conducted under similar
conditions with the 1 ml reaction mixture containing: 700 llof
receptor preparation; 100 ul of either binding buffer (for total
binding) or 10 lM mianserin (final concentration for non-specific
binding); 100 ll of the tritiated agent (final concentration of
0.2 nM [3H] ketanserin); and 100 ll of diluted CBD or binding
buffer in case of controls.
cAMP Assay. CHO cells were cultured to confluency in 12-
or 24-well plates (27). Medium was aspirated and the cells were
rinsed twice in warm, serum-free F-12 medium. Cells were then
incubated for 20 min. at 37°C in 0.5 mls of serum-free F-12 med-
ium containing 100 lM isobutylmethylxanthine (IBMX) and the
following substances (final concentrations) alone or in combination
(see Fig. 3): 30 lM forskolin (FSK; for all treatments); 1 lM5-
HT; 16 lM CBD; and 0.05 lM NAN-190 (NAN). Reactions were
stopped by aspiration of medium and addition of 0.5 ml of
100 mM HCl. After 10 min., well contents were removed and
centrifuged at 4000 rpm. Supernatants were diluted in 100 mM
HCl, and cAMP was quantified (27) directly in a microplate format
by colorimetric enzyme immunoassay (EIA) with a kit from Assay
Designs (Ann Arbor). Triplicate independent samples were assayed
in quadruplicate to increase precision.
[35S]GTPcS Assay. H5-HT1aR membranes from trans-
fected CHO cells were incubated with 5-HT (0.1 lM) and/or CBD
1038 Russo, Burnett, Hall, and Parker
(16 lM); see Fig.2), and the following incubation mixture: 20 mM
HEPES buffer, pH 7.4, 5 mM MgCl2, 1 mM EDTA, 1 mM DTT,
100 mM NaCl, 100 uM GDP, 10 lM pargyline, 0.2 mM ascor-
bate, and 0.1 nM [35S]GTPcS (32). Mixtures were incubated for
30 min. at 30°C, and were terminated by dilution in cold buffer.
The mixture was filtered on GF/C filters, rinsed twice in buffer,
followed by drying and liquid scintillation counting. Negative
control (basal incorporation) was the above mixture minus CBD or
5-HT. Non-specific binding was determined in the presence of cold
GTPcS-(10 lM). Positive control was H5-HT1aR membranes in
the same incubation mixture plus 5-HT. All values reported in
Fig. 2 are for specific binding (total non-specific) of triplicates.
Dilution of Cannabinoids. CBD and THC were obtained in
dilute (1 mg/ml) solution from Sigma Chemical Co. (St. Louis,
MO). These solutions were stored at 4°C until use and then diluted
in distilled water and finally in the buffer appropriate to the par-
ticular assay. Fresh dilutions of cannabinoids were made daily.
Each final concentration of cannabinoid thus contained some of
the vehicle (methanol). The highest concentration of methanol
encountered in any assay (1%) was then tested in that assay system
for activity. In pair-wise comparative t testing, none of the meth-
anol controls were found to be distinguishable from negative
control (buffer).
Statistical Analysis. All statistics (means, standard devia-
tions, standard errors of the mean (SEM), and t tests) were per-
formed with software provided by Poly Software International; in
some cases, statistics were corroborated by hand using a Hewlett-
Packard Graphing Calculator, HP48. Graphs were constructed
with Excel software provided by Microsoft.
RESULTS
Cannabidiol produces concentration-dependent
displacement of the agonist [3H[8-OH-DPAT from
the H5-HT1aR (Fig. 1). Using crude membrane
preparations from cultured CHO cells transfected
with H5-HT1aR (See methods), CBD diluted in
methanolic buffer displaced agonist by 73 ± 8 %
(S.E.M.) at 16 lM. The highest concentration of
methanol (1%) present at 32 lM CBD produced
only 3 ± 0.5% displacement of agonist, a level
indistinguishable from control when the methanol
and control means are compared statistically. While
CBD was active in the micromolar range, tetrahy-
drocannabinol was unable (108 ± 6% of control) to
produce agonist displacement at a concentration of
32 lM.
The ability of CBD to produce concentration-
dependent displacement of highly potent and specific
agonist from the H5-HT1aR ligand-binding site
raised the question of the intrinsic activity of CBD.
Experiments were designed to test the agonistic
potential of cannabidiol. Since H5-HT1aR is G pro-
tein-coupled, agonist binding would be expected to
increase GTP binding, measurable when the stable
analog of GTP, GTP cS is present in a radiolabeled
form. 0.1 lM 5HT increased [35S]GTPcS incorpora-
tion by 57 ± 7% (Fig. 2) above the basal level (buffer)
in membranes of CHO transfected with the receptor.
Similarly, 16 lM CBD increased [35S]GTPcS incor-
poration by 67 ± 6% above the basal level. Together,
5-HT and CBD increased [35S]GTPcS incorporation
to 123 ± 10% above the basal level, suggesting that
CBD had not reached its maximum possible stimu-
Fig. 1. Displacement of Specifically-Bound [3H]8-OH-DPAT By Cannabidiol (CBD) and Tetrahydrocannabinol (THC) In Membranes
Containing the Human 5-HT1a Receptor. Concentrations are micromolar. Values are the mean ± SEM with n’s=3–6. More detailed
experimental conditions of cell culture, membrane preparation, and drug-receptor binding are outlined in Experimental Procedure.
5-HT1a Receptor Agon ism by Cannabidiol 1039
lation. By reference to CBD’s displacement capacity
at the receptor’s ligand binding site (Fig. 1), 16 lM
CBD occupies about 73% of the available binding
sites.
To further test the hypothesis that CBD is an
agonist at H5-HT1aR, experiments were designed to
measure activity in the adenylyl cyclase (AC) system
negatively coupled to the receptor. In this format, AC
is first stimulated by the natural product forskolin
(FSK) at a concentration of 30 lM (control = 100 ±
5%). 1 lM of the agonist 5-HT reduced FSK -stimu-
lated cAMP to 29 ± 8% of control (Fig. 3). 16 lM
CBD reduced FSK-stimulated cAMP to 38 ± 3% of
control. At a concentration of 0.05 lM, the highly
specific 5-HT1aR antagonist NAN-190 reduced the
5-HT effect to 60 ± 7% of control and the CBD effect
to 76 ± 5% of control, providing further evidence
that CBD is acti ng at the ligand- binding site of H5-
0
50
100
150
200
250
Control 5HT (0.1) CBD (16) 5HT/CBD
% Control (Specific [35S]γ-S-GTP Incorp.)
*
**
Fig. 2. Incorporation of [35S]GTPcS by Cannabidiol (CBD) In Membranes Containing the Human 5-HT1a Receptor. Control represents
incorporation in the basal setting (buffer). Concentrations in micromolar are: 5-HT (0.1); CBD (16). Results are expressed relative to basal
incorporation as mean ± SEM with n’s=9–18. *P<0.01, relative to Control;**P<0.01, relative to 5HT. Further experimental details are
found in Experimental Procedure.
Fig. 3. Inhibition of Forskolin (FSK)-Stimulated cAMP by Cannabidiol (CBD), Serotonin (5-HT), and the inhibitor NAN-190 (NAN) in
Whole Cells Transfected With the Human 5-HT1a Receptor. All conditions contain FSK at 30 lM and the phosphodiesterse inhibitor
isobutylmethylxanthine (IBMX) at 100 lM. Other concentrations in micromolar are: 5-HT (1); CBD (16); and NAN (0.05). Results are
expressed as percentage of FSK control as mean ± SEM with n’s = 3–6. *P<0.05, relative to 5-HT; **P<0.01, relative to CBD. Further
experimental details are found in Experimental Procedure.
1040 Russo, Burnett, Hall, and Parker
HT1aR. At the concentration used here (0.05 lM),
NAN-190 does not reduce FSK-stimulated cAMP
levels on its own (data not shown).
Since the 5-HT2aR is another receptor puta-
tively involved in the pathogenesis of migraine
headache, we conducted a limited comparison at
cloned rat 5-HT2aR. At the highest concentration
of CBD tested (32 lM), 50 ± 5% of [3H]Ketans-
erin is displaced from membrane preparations of
the cloned rat 5-HT2aR. The displacement is con-
centration-dependent as lower concentrations of
CBD progressively displace less ketanserin, until at
8 lM CBD, the effect is barely above control level.
Comparatively, then, CBD is less potent in dis-
placement from the rat 5-HT2aR relative to H5-
HT1aR. As with H5-HT1aR, THC (32 lM) is
inactive in displacement from rat 5-HT2aR. Signal
transduction properties of CBD at rat 5-HT2aR
have not been explored yet.
DISCUSSION
There is substantial literat ure to support the idea
that tetrahydrocannabinol (THC) is responsible for
many of the meaningful and diverse components of
cannabis’ pharmacological activity (33), but other
available evidence supports important contributions
of CBD and other phytocannabinoids and terpenoids
to its pharmacological activity (34,35). It is well
established that the pharmacology of cannabis
combines therapeutic properties (e.g., benefits on
neuropathic pain and spastici ty) (36–39), and lower
urinary tract symptoms (40) that must be weighed
against adverse effects such as intoxication that may
be counter-productive in a therapeutic sense. A
prominent example of the latter is the hallucinogenic
potential of cannabis demonstrated at higher doses,
especially in certain cultural settings. There is also an
outstanding body of experimental evidence to suggest
that THC is hallucinogenic while the closely related
cannabinoid, cannabidiol (CBD) opposes such
activity (3,41).
In pursuit of those pharmacologic al actions of
cannabis that may underlie some of its medicinally
important possibilities, differentiation between TH C
and CBD at the receptor level may be of significance.
This could be especially so at non-cannabinoid
receptors such as 5-HT recept ors. The results shown
in Fig. 1 establish such a contrast in that CBD shows
micromolar affinity in displacing a known agonist,
[3H]8-OH-DPAT, from the 5-HT1aR ligand-binding
site, THC is inactive in the same concentration
range.
CBD’s 5-HT1aR potency could underlie activity
anywhere along the intrinsic activity continuum from
full agonist to silent antagonist. Experiments sum-
marized in Figs. 2 and 3 provide evidence that CBD
is likely to behave a s an agonist in this receptor sys-
tem. Thus, CBD demonstrated the ability to increase
GTP binding to the receptor coupled G protein, Gi,
which is characteristic behaviour of a receptor ago-
nist. These GPCR are further linked to effector signal
transduction sub-systems such as, in the case of a Gi
GPCR, the AC step in cAMP regulation. In Fig. 3,
when AC is stimulated by forskolin (FSK), the ago-
nist 5-HT markedly reduces cAMP production in this
negatively coupled complex. Likewise, CBD acts as
an agonist in these experiments by reducing cAMP
concentration. The results in Figs. 2 and 3 together
support the hypothesis that CBD is an agonist.
Although not completely conclusive in demonstrating
whether CBD is a full or partial agonist, the com-
parable power of CBD and 5-HT at concentrations
that represent less than full receptor occupancy
(Fig. 1) lend support to the full agonist concept.
The contrast between CBD and THC regarding
their interactions at 5-HT1aR relative to CB1R is
striking. THC is at least 10 times more potent in
binding to CB1R; at 5-HT1aR the relationship is just
the opposite, where CBD has micromolar affinity,
and THC shows no binding in the micromo lar range.
At CB1R, THC has sub-micromolar affinity, yet
CBD has micr omolar affinity. The comparison con-
tinues into the realm of signal transduction, where at
CB1R, CBD is putatively an antagonist or inverse
agonist (2); at 5-HT1aR, we have concluded that
CBD is an agonist.
What implications do these results at 5-HT1aR
have for CBD and cannabis? Cannabis is a very
complex mixture of chemical compounds (42), as is
true of most crude natural product drug mixtures.
The dearth of biochemical investigations with non-
psychoactive cannabis components, such as CBD,
create a void of understanding regarding the use of
one or more of these pharmacologically active com-
ponents as therapeutic agents. It has recently been
demonstrated that CBD stimulates TRPV1 (one of
the vanilloid receptors), inhibits the reuptake of
anandamide, and weakly inhibits its hydrolysis (42),
thus making it possibly the first pharmacotherapeutic
agent to modulate endocannabinoid function (1). As
anandamide has already shown activity at 5-HT1aR,
and 36% inhibition of function at 5-HT2aR (14), the
5-HT1a Receptor Agon ism by Cannabidiol 1041
psychopharmacological importance of such relation-
ships is underscored.
The results reported here argue that CBD is active
as an agonist in vitro at H5-HT1a R and that CBD may
also have in vitro actions at the rat 5-HT2aR. Should
CBD prove to have antagonistic activity at 5-HT2A, it
would support its role as a migraine prophylactic agent
(19). Together, these results lend credence to the idea
that CBD and related compounds merit study at a
variety of receptor systems, in a number of species, and
at various levels from the molecular to whole animal.
If, for example, CBD demonstrates clinical activity at
5-HT1aR in vivo, therapeutic possibilities could arise
in a variety of neurological and other physiologically
relevant settings.
ACKNOWLEDGMENTS
The authors would like to thank the following individuals for
their assistance and ideas that contributed to this project: Rustem
Medora, Alicia Christians, Cortney Halley, Sonja Sakaske, Ben
Seaver, and Lynn Parker . The following agencies are gratefully
acknowledged for their financial support of the work: NIH NIG-
MS grants #: GM/OD 54302–01 and 02 and NIH NCRR grant #:
P20 RR 15583 to the NIH COBRE Center for Structural and
Functional Neuroscience.
REFERENCES
1. Russo, E. B. 2003. Introduction: Cannabis: from pariah to
prescription. J. Cann. Therap. 3(3–4):1–29.
2. Pertwee, R. G. 2004. The pharmacology and therapeutic po-
tential of Cannibidiol. Pages 1–52, in DiMarzo, V. (ed.),
Cannabinoids. Kluwer Academic/Plenum Publishers.
3. Zuardi, A. W. and Guimaraes, F. S. 1997. Cannabidiol as an
anxiolytic and antipsychotic. Pages 133–141, in Mathre, M. L.
(ed.), Cannabis in medical practice: a legal, historical and
pharmacological overview of the therapeutic use of marijuana.
NC: McFarland: Jefferson.
4. Carlini, E. A. and Cunha, J. M. 1981. Hypnotic and antiepi-
leptic effects of cannabidiol. J. Clin. Pharmacol. 21(8–9 Sup-
pl):417S–427S.
5. Hampson, A. J., Grimaldi, M., Axelrod, J., and Wink, D.
1998. Cannabidiol, (-)Delta9-tetrahydrocannabinol are neuro-
protective antioxidants. Proc. Natl. Acad. Sci. USA.
95(14):8268–73.
6. Iuvone, T., Esposito, G., Esposito, R., Santamaria, R., Di
Rosa, M., and Izzo, A. A. 2004. Neuroprotective effect of
cannabidiol, a non-psychoactive component from cannabis
sativa, on beta-amyloid-induced toxicity in PC 12 cells. J.
Neurochem. 89:134–141.
7. Nicholson, A. N., Turner, C., Stone, B. M., and Robson, P. J.
2004. Effect of delta-9-tetrahydrocannabinol and cannabidiol
on nocturnal sleep and early-morning behavior in young
adults. J. Clin. Psychopharmacol. 24(3):305–313.
8. McPartland, J., Di Marzo, V., De Petrocellis, L., Mercer, A.,
and Glass, M. 2001. Cannabinoid receptors are absent in in-
sects. J. Comp. Neurol. 436(4):423–9.
9. Pertwee, R.G. 1997. ‘‘Pharmacology of cannabinoid CB1 and
CB2 Receptors’’. Pharmacol. Therap. 74(2):129–180.
10. Thomas, B. F., Gilliam, A. F., Burch, D. F., Roche, M. J., and
Seltzman, H. H. 1998. ‘‘Comparative receptor binding analysis
of cannabinoid agonists and antagonists’’. J. Pharm. Exp.
Therap 285(1):285–292.
11. Petitet, F., Jeantaud, B., Reibaud, M., Imperato, A., and
Dubroeueq, M. C. 1998. ‘‘Complex pharmacology of natural
cannabinoids: Evidence for partial agonist activity at delta 9-
tetrahydrocannabinol and antagonist activity of cannabidiol
on rat brain cannabinoid receptors’’. Life Sci. 63 (1):PL1–PL6.
12. McPartland, J. M. and Russo, E. B. 2001. Cannabis and
Cannabis Extracts: Greater Than the Sum of Their Parts? J.
Cannabis Therap. 1(3–4):103–132.
13. Russo, E. B. 2001. Hemp for headaches: An in-depth historical
and scientific review of cannabis in migraine treatment. J.
Cannabis Therap. 1(2):21–92.
14. Russo, E. B. 2004. Clinical endocannabinoid deficiency
(CECD): Can this concept explain therapeutic benefits of
cannabis in migraine, fibromyalgia, irritable bowel syndrome
and other treatment-resistant conditions? Neuroendocrinol.
Lett. 25(1–2):31–39.
15. Ferrari, M. D. and Saxena, P. R. 1995. 5-HT1 receptors in
migraine pathophysiology and treatment. Eur. J. Neurol.
2:5–21.
16. Hershkowitz, M. 1978. ‘‘The effect of in vivo treatment with (-)
/delta 1-tetrahydrocannabinol, and other psychoactive drugs
on the in vitro uptake of biogenic amines’’. Adv. Biosci.
22–23:351–358.
17. Cowen, P. J. 2000. Psychopharmacology of 5-HT1a receptors.
Nuc. Med. Biol. 27:437–439.
18. Barnes, N. M. and Sharp, T. 1999. A review of central 5-HT
receptors and their function. Neuropharmacol. 38(8):1083–
1152.
19. Fargin, A., Raymond, J. R., Lohse, M. J., Kobilka, B. K.,
Caron, M. G., and Lefkowitz, R. J. 1988. The genomic clone
G-21 which resembles a beta-adrenergic receptor sequence
encodes the 5-HT1a receptor. Nature 335:358–360.
20. Nelson, D. L., Monroe, P. J., Lambert, G., and Yamamura, H.
I. 1987. [3H]Spiroxatrine labels a serotonin 1a-like site in rat
hippocampus’’. Life Sci. 41:1567–1576.
21. Weber, J. T., O’Connor, M.-F., Hayataka, K., Colson, N.,
Medora, R., Russo, E. B., and Parker, K. K. 1997. Activity of
parthenolide at 5HT2a receptors. J. Nat. Prods. 60(6):651–653.
22. Glennon, R. A., Teitler, M., and Sanders-Bush, E. 1992.
Hallucinogens and serotonergic mechanisms. NIDA Res.
Monographs 119:131–135.
23. Hedlund, P. B., Carson, M. J., Sutcliffe, J. G., and Thomas, E.
A. 1999. Allosteric regulation by oleamide of the binding
properties of 5-hydroxytryptamine 7 receptors. Biochem.
Pharmacol. 58:1807–1813.
24. Boger, D. L., Patterson, J. E., and Jin, Q. 1998. Structural
requirements for 5-HT2A and 5-HT1A serotonin receptor
potentiation by the biologically active lipid oleamide. Proc.
Natl. Acad, Sci, USA 95(8):4102–4107.
25. Cheer, J. F., Cadogan, A. K., Marsden, C. A., Fone, K. C.,
and Kendall, D. A. 1999. ‘‘Modification of 5HT2a receptor
1042 Russo, Burnett, Hall, and Parker
mediated behaviour in the rat by oleamide and the role of
cannabinoid receptors’’. Neuropharmacol. 38(4):533–545.
26. Devlin, M. G. and Christopoulos, A. 2002. ‘‘Modulation of
cannabinoid agonist binding by 5-HT in the rat cerebellum’’. J.
Neurochem. 80(6):1095–1102.
27. Ortiz, T. C., Devereaux, M. C., and Parker, K. K. 2000.
Structural variants of a human 5-HT1a receptor intracellular
loop 3 peptide. Pharmacol. 60:195–202.
28. Julius, D., Huang, K. N., Livelli, T. J., Axel, R., and Jessel, T.
M. 1990. The 5HT2 receptor defines a family of structurally
distinct but functionally conserved serotonin receptors’’. Proc.
Natl. Acad. Sci.USA 87:928–932.
29. Hall, B., Burnett, A., Halley, C., Parker, L., Russo, E., and
Parker, K. K. 2004. Pharmacology of cannabidiol at serotonin
receptors. Proc. West. Pharmacol. Soc. 47:43–(M24).
30. Pierce, P. A. and Peroutka, S. J. 1989. Hallucinogenic drug
interactions with neurotransmitter receptor binding sites in
human cortex’’. Psychopharmacol. 97:118–122.
31. Bradford, M. M. 1976. A rapid sensitive method for the
quantitation of microgram quantities of protein utilizing the
principle of protein-dye binding. Anal. Biochem. 72:248–254.
32. Wieland, T. and Jacobs, K. H. 1994. Measurement of receptor-
stimulated guanosine 5’-O-(gamma-thio)triphosphate)binding
by G proteins. Meth. Enzymol. 237:3–27.
33. Wachtel, S. R., ElSohly, M. A., Ross, R. A., Ambre, J., and de
Wit, H. 2002. Comparison of the subjective effects of delta9-
tetrahydrocannabinol and marijuana in humans. Psychophar-
macology 161:331–339.
34. McPartland, J. M. and Russo, E. B. 2001. Cannabis and
cannabis extracts: Greater than the sum of their parts? J. Cann.
Therap. 1(3–4):103–132.
35. Russo, E. B. and McPartland, J. M. 2003. Cannabis is more
than simply Delta(9)-tetrahydrocannabinol. Psychopharma-
cology (Berl.) 165(4):431–2.
36. Berman, J. S., Symonds, C., and Birch, R. 2004. Efficacy of two
cannabis based medicinal extracts for relief of central neuro-
pathic pain from brachial plexus avulsion: results of a ran-
domized controlled trial. Pain 112(3):299–306.
37. Notcutt, W., Price, M., Miller, R., Newport, S., Phillips, C.,
Simmonds, S., and Sansome, C. 2004. Initial experiences with
medicinal extracts of cannabis for chronic pain: results from 34
‘‘N of 1’’ studies. Anaesthesia 59:440–452.
38. Wade, D. T., Makela, P., Robson, P., House, H., and Bat-
eman, C. 2004. Do cannabis-based medicinal extracts have
general or specific effects on symptoms in multiple sclerosis? A
double-blind, randomized, placebo-controlled study on 160
patients. Mult. Scler. 10(4):434–41.
39. Wade, D. T., Robson, P., House, H., Makela, P., and Aram, J.
2003. A preliminary controlled study to determine whether
whole-plant cannabis extracts can improve intractable neuro-
genic symptoms. Clin. Rehabil. 17:18–26.
40. Brady, C. M., DasGupta, R., Dalton, C., Wiseman, O. J.,
Berkley, K. J., and Fowler, C. J. 2004. An open-label pilot
study of cannabis based extracts for bladder dysfunction in
advanced multiple sclerosis. Mult. Scler. 10:425–433.
41. Karniol, I. G., Shirakana, I., Kasinski, N., Pfeferman, A., and
Carlini, E. A. 1974. ‘‘Cannabidiol Interferes With the Effects of
Delta 9-Tetrahydrocannabinol in Man’’. Eur. J. Pharmacol.
28(1):172–177.
42. Bisogno, T., Hanus, L., De Petrocellis, L., Tchilibon, S.,
Ponde, D. E., Brandi, I., Moriello, A. S., Davis, J. B., Mech-
loulam, R., and DiMarzo, V. 2001. Molecular targets for
cannabidiol and its synthetic analogues: effect on vanilloid
VR1 receptors and on the cellular uptake and enzymatic
hydrolysis of anandamide. Br. J. Pharmacol. 134(4):845–852.
43. Howlett, A.C. 2002. ‘‘International union of pharmacology
XXVII classification of cannabinoid receptors’’. Pharmacol.
Rev. 54(2):161–202.
5-HT1a Receptor Agon ism by Cannabidiol 1043
... These disorders may have a long-term or recurrent course and therefore contribute to the alteration of the quality of life of PD patients [105]. CBD is hypothesized to affect depression due to its capacity to target and modulate serotonin and norepinephrine brain neurotransmission as well as its active binding to 5HT-1A receptors [106]. Furthermore, CBD promotes synaptic plasticity and neurogenesis, both of which are important in the development and treatment of depression [21,52,106]. ...
... CBD is hypothesized to affect depression due to its capacity to target and modulate serotonin and norepinephrine brain neurotransmission as well as its active binding to 5HT-1A receptors [106]. Furthermore, CBD promotes synaptic plasticity and neurogenesis, both of which are important in the development and treatment of depression [21,52,106]. The conclusions of certain animal models have been positive and encouraging [107,108]. ...
Article
Full-text available
Background Parkinson’s disease (PD) is primarily known as a motor disorder; however, its debilitating non-motor symptoms have a significant impact on patients’ quality of life. The current standard treatment, l-DOPA, is used to relieve motor symptoms, but prolonged use is often associated with severe side effects. This creates an urgent need for effective alternatives targeting both motor and non-motor symptoms. Objectives Over the past decade, Cannabis sativa and its cannabinoids have been widely studied across various health conditions. Among these compounds, cannabidiol (CBD), a non-psychoactive component, is garnering growing interest due to its multi-targeted pleiotropic properties. This work aims to provide a comprehensive overview of CBD’s efficacy in PD. Methods This review compiles data on both motor and non-motor symptoms of PD, integrating results from preclinical animal studies and available clinical trials. Results Preclinical research has demonstrated promising results regarding CBD’s potential benefits in PD; however, the total number of clinical trials is limited (with only seven studies to date), making it difficult to draw definitive conclusions on its efficacy. Conclusions While preclinical findings suggest that CBD may have therapeutic potential in PD, the limited number of clinical trials highlights the need for further research. This review emphasizes the gaps that need to be addressed in future studies to fully understand CBD’s role in treating both motor and non-motor symptoms of PD.
... By itself, CBD did not interfere with short-and long-term memory, corroborating findings in the water maze Morris test [107]. Although initially described as a 5-HT1A full agonist [108], CBD probably acts as a positive allosteric modulator of these receptors [109]. The involvement of serotonergic pathways in CBD-mediated effects corroborates results observed with atypical antipsychotics, such as aripiprazole, clozapine, lurasidone, tandospirone, and ziprasidone, which also act as partial agonists of 5-HT1A receptors [110][111][112]. ...
... It is made available under a preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in The copyright holder for this this version posted October 22, 2024. ; https://doi.org/10.1101/2024.10.21.619352 doi: bioRxiv preprint by facilitating the activation of 5-HT1A receptors expressed, among others, on PV + interneurons [108]. In the GABAergic interneurons of the PFC and hippocampus, a high density of 5-HT1A receptors is responsible for neuronal hyperpolarization [121] and consequent disinhibition of pyramidal neurons in the hippocampus, which in turn regulate γ oscillations [122]. ...
Preprint
Full-text available
Cannabidiol (CBD) has shown potential for treating schizophrenia (SCZ) by targeting its positive, negative, and cognitive symptoms. In this study, we investigated if CBD could reverse the memory impairment observed after chronic administration of the NMDA receptor antagonist. MK-801 treatment (0.5 mg/kg i.p., twice a day, for 14 days) resulted in short- and long-term memory deficits and decreased relative power of γ oscillation in freely moving animals. CBD administration (60 mg/kg i.p. daily for seven days after the MK-801 treatment period) reversed these changes. The positive cognitive effects of CBD were prevented by a 5-HT1A, but not a CB2, receptor antagonist. On the cellular level, CBD reversed MK-801-induced reduced number of parvalbumin-positive neurons and their associated perineuronal nets in the prelimbic medial prefrontal cortex (mPFC) and ventral hippocampus (vHip). This neuroprotective effect was mediated by 5-HT1A and CB2 receptors in the vHip but was independent of these receptors in the mPFC. Additionally, CBD reversed MK-801-induced microglial activation in both mPFC and vHip, again through 5-HT1A and CB2 receptors. These findings suggest that CBD modulates multiple pathways affected in SCZ-like conditions, offering a promising therapeutic avenue for SCZ treatment. Chemical compounds studied in this article CBD (PubChem CID: 644019), MK-801 (PubChem CID: 180081), AM630 (PubChem CID: 4302963), WAY100635 (PubChem CID: 11957721).
... One primary mechanism is through its interaction with the endocannabinoid system, particularly by enhancing the signaling of anandamide, an endogenous cannabinoid, which contributes to mood regulation and stress resilience [22]. Additionally, CBD acts as an agonist at serotonin 5-HT 1A receptors, a crucial pathway involved in anxiety and mood disorders [23]. CBD also exerts influence on the hypothalamic-pituitary-adrenal axis, reducing the release of stress hormones such as cortisol [21]. ...
Article
Full-text available
In recent years, the European market, including Romania, has witnessed a significant increase in the promotion of cannabidiol (CBD)-based products, often presented as effective treatments for various health conditions. This study investigates the inconsistencies between the health claims associated with these supplements and the evidence from clinical trials. To identify products available on the Romanian market, a systematic review of online pharmacies and websites that specialize in selling CBD-based products has been performed. Additionally, a systematic review of clinical trials has been conducted to assess the efficacy of CBD for the specified indications. Our analysis revealed that some claims, such as those related to post-traumatic stress disorder, lack substantial clinical evidence. Moreover, even when clinical support exists, the dosages recommended for the supplements are often significantly lower than those used in trials, raising concerns about their efficacy. These findings highlight the need for stricter regulatory oversight and more transparent communication to ensure that consumer expectations are aligned with scientific evidence, ultimately promoting informed decision-making and consumer safety.
... Cannabidiol has low binding affinities for CB1 and CB2 receptor but has been hypothesized to act on the endocannabinoid system as a FAAH inhibitor [101], a negative allosteric modulator of the CB1 receptor [102], and an inverse agonist of the CB2 receptor [103]. Other proposed mechanisms by which it may have therapeutic effects specifically within the context of CUD and other substance use disorders include its modulatory actions on glutamate-GABA systems [104] and its potential 5HT1A agonist effects [105]. Preclinical studies using alcohol and cocaine models of substance use disorder demonstrated that CBD decreased drug self-administration [106,107]. ...
Article
Full-text available
With rapid expansion of cannabis legalization worldwide, rates of cannabis use and cannabis use disorder (CUD) are increasing; the need for safe and effective medications to treat CUD is urgent. This narrative review evaluates evidence for promising pharmacotherapies to treat CUD from randomized, placebo-controlled trials. Pharmacotherapies for CUD are categorized based on compound targets (e.g., cannabinoid receptor 1 [CB1] agonists such as nabilone, serotonergic compounds such as bupropion, GABAergic compounds such as zolpidem) and outcomes are organized by predetermined withdrawal symptoms, cannabis craving, and cannabis relapse/use. Most promising pharmacotherapies for CUD are drugs that act on the endocannabinoid system and specifically at the CB1 receptor. Priority populations such as females, certain racial/ethnic groups, and age groups experience a different course of CUD progression, symptoms, and drug effects that are important to consider when evaluating outcomes related to CUD. Possible explanations for these disparities are explored, along with the clinical trials that explore these demographics in treating CUD with pharmacotherapies.
... Unlike THC, CBD does not exhibit a high affinity at the CB1 and CB2 receptors but can function as a negative allosteric modulator of CB1 [43], indirectly influencing endocannabinoid signaling. Additionally, CBD acts as an agonist at the 5-HT1A serotonin receptor [44], contributing to its anxiolytic and antidepressant effects. It also modulates TRPV1/2 [45], which are involved in inflammation and pain perception. ...
Preprint
Full-text available
Cannabis sativa is known for producing over 120 distinct phytocannabinoids, with Δ9-tetrahydrocannabinol (Δ9-THC) and cannabidiol (CBD) being the most prominent, primarily in their acidic forms. Beyond Δ9-THC and CBD, a wide array of lesser-known phytocannabinoids, along with terpenes, flavonoids, and alkaloids, demonstrate diverse pharmacological activities, interacting with the endocannabinoid system (eCB) and other biological pathways. These com-pounds, characterized by phenolic structures and hydroxyl groups, possess lipophilic properties, allowing them to cross the blood-brain barrier (BBB) effectively. Notably, their antioxidant, an-ti-inflammatory, and neuro-modulatory effects position them as promising agents in treating neu-rodegenerative disorders. While research has extensively examined the neuropsychiatric and neuroprotective effects of Δ9-THC, other minor phytocannabinoids remain underexplored. Given the well-established neuroprotective potential of CBD, there is growing interest in the therapeutic benefits of non-psychotropic minor phytocannabinoids (NMPs) in brain disorders. This review highlights the emerging research on these lesser-known compounds and their neuroprotective potential. It offers insights into their therapeutic applications across various major neurological conditions.
... 5-HT1a receptors are expressed as both pre-synaptic and post-synaptic receptors within the brain, making them relevant in treating psychiatric disorders [24]. CBD was found to have moderate affinity and display agonist activity at 5-HT1a receptors [25]. The agonistic activity of CBD at 5-HT1a has been suggested as the basis behind the anxiolytic effect of CBD [26]. ...
Article
Full-text available
Cannabidiol (CBD) is a major non-psychotropic phytocannabinoid that exists in the Cannabis sativa plant. CBD has been found to act on various receptors, including both cannabinoid and non-cannabinoid receptors. In addition, CBD has antioxidant effects that are independent of receptors. CBD has demonstrated modulatory effects at different organ systems, such as the central nervous system, immune system, and the gastrointestinal system. Due to its broad effects within the body and its safety profile, CBD has become a topic of therapeutic interest. This literature review summarizes previous research findings with regard to the effect of CBD on the gastrointestinal (GI) system, including its effects at the molecular, cellular, organ, and whole-body levels. Both pre-clinical animal studies and human clinical trials are reviewed. The results of the studies included in this literature review suggest that CBD has significant impact on intestinal permeability, the microbiome, immune cells and cytokines. As a result, CBD has been shown to have therapeutic potential for GI disorders such as inflammatory bowel disease (IBD). Furthermore, through interactions with the gut, CBD may also be helpful in the treatment of disorders outside the GI system, such as non-alcoholic liver disease, postmenopausal disorders, epilepsy, and multiple sclerosis. In the future, more mechanistic studies are warranted to elucidate the detailed mechanisms of action of CBD in the gut. In addition, more well-designed clinical trials are needed to explore the full therapeutic potential of CBD on and through the gut.
Article
Full-text available
In Europe, industrial hemp (Cannabis sativa L.) is currently cultivated primarily for its fibers and seeds due to regulatory restrictions on flower use, although the flowers present untapped valorization potential. This study assesses the economic viability of cultivating hemp for its inflorescences and examines the quality impact of different cultivation methods by testing two varieties, Santhica 27 and Félina 32, across distinct growth environments. The cultivation systems tested include greenhouse-based soilless setup and open-field production. Within the greenhouse, the effects of an ethylene-based growth regulator, applied during the vegetative stage, on plant morphology and cannabinoid concentrations were assessed at three dosage levels (7.5, 15, and 30 µL L−1). The results showed a dose-dependent reduction in shoot height, lasting 21 to 28 days post-treatment before diminishing. Comparable total cannabigerol (CBG) levels were achieved with Santhica 27 in both field conditions and at the 30 µL L−1 ethylene dose in the greenhouse, while total cannabidiol (CBD) levels in Félina 32 were higher in the field than in the greenhouse. Overall, greenhouse cultivation yielded significantly greater cannabinoid production per square meter due to increased inflorescence biomass and the potential for multiple harvest cycles. However, operating costs for greenhouse cultivation are at least 13 to 15 times higher than those of open-field production. Economic analysis suggests that optimizing greenhouse cultivation techniques and reducing labor requirements at harvest could help mitigate these costs, enhancing the economic feasibility of greenhouse-based hemp flower production.
Article
Full-text available
Cannabis sativa is known for producing over 120 distinct phytocannabinoids, with Δ9-tetrahydrocannabinol (Δ9-THC) and cannabidiol (CBD) being the most prominent, primarily in their acidic forms. Beyond Δ9-THC and CBD, a wide array of lesser-known phytocannabinoids, along with terpenes, flavonoids, and alkaloids, demonstrate diverse pharmacological activities, interacting with the endocannabinoid system (eCB) and other biological pathways. These compounds, characterized by phenolic structures and hydroxyl groups, possess lipophilic properties, allowing them to cross the blood–brain barrier (BBB) effectively. Notably, their antioxidant, anti-inflammatory, and neuro-modulatory effects position them as promising agents in treating neurodegenerative disorders. While research has extensively examined the neuropsychiatric and neuroprotective effects of Δ9-THC, other minor phytocannabinoids remain underexplored. Due to the well-established neuroprotective potential of CBD, there is growing interest in the therapeutic benefits of non-psychotropic minor phytocannabinoids (NMPs) in brain disorders. This review highlights the emerging research on these lesser-known compounds and their neuroprotective potential. It offers insights into their therapeutic applications across various major neurological conditions.
Article
Full-text available
OBJECTIVES: This study examines the concept of clinical endocannabinoid deficiency (CECD), and the prospect that it could underlie the pathophysiology of migraine, fibromyalgia, irritable bowel syndrome, and other functional conditions alleviated by clinical cannabis. METHODS: Available literature was reviewed, and literature searches pursued via the National Library of Medicine database and other resources. RESULTS: Migraine has numerous relationships to endocannabinoid function. Anandamide (AEA) potentiates 5-HT1A and inhibits 5-HT2A receptors supporting therapeutic efficacy in acute and preventive migraine treatment. Cannabinoids also demonstrate dopamine-blocking and anti-inflammatory effects. AEA is tonically active in the periaqueductal gray matter, a migraine generator. THC modulates glutamatergic neurotransmission via NMDA receptors. Fibromyalgia is now conceived as a central sensitization state with secondary hyperalgesia. Cannabinoids have similarly demonstrated the ability to block spinal, peripheral and gastrointestinal mechanisms that promote pain in headache, fibromyalgia, IBS and related disorders. The past and potential clinical utility of cannabis-based medicines in their treatment is discussed, as are further suggestions for experimental investigation of CECD via CSF examination and neuro-imaging. CONCLUSION: Migraine, fibromyalgia, IBS and related conditions display common clinical, biochemical and pathophysiological patterns that suggest an underlying clinical endocannabinoid deficiency that may be suitably treated with cannabinoid medicines.
Article
Full-text available
Cannabis has been employed in human medicine for more than 4000 years. In the last century, political prohibition led to its disappearance from the conventional pharmacopoeia, but this trend is reversing due to the broad acceptance and application of this forbidden medicine by patients with chronic and intractable disorders inadequately treated by available therapeutics. This study addresses the “road back” for cannabis medicines, and reacceptance as prescription products.Current pharmacology of the two primary therapeutic phytocannabinoids, THC and CBD, is reviewed with respect to herbal synergy and as pertains to treatment of pain, spasm and the wide range of therapeutic applications and adverse effects of cannabis.In particular, the efforts of GW Pharmaceuticals to develop cannabis based medicine extracts (CBME) are documented including cultivation of genetically-selected medical-grade cannabis cloned strains in glass houses with organic and integrated pest management techniques, and their processing employing supercritical carbon dioxide extraction and winterization. These CBMEs are then available for formulation of dosage forms including sublingual extracts and inhaled forms. An optional Advanced Delivery System (ADS) is also discussed.
Article
Full-text available
. A central tenet underlying the use of botanical remedies is that herbs contain many active ingredients. Primary active ingredients may be enhanced by secondary compounds, which act in beneficial syn-ergy. Other herbal constituents may mitigate the side effects of dominant active ingredients. We reviewed the literature concerning medical can-nabis and its primary active ingredient, ∆ 9 -tetrahydrocannabinol (THC). Good evidence shows that secondary compounds in cannabis may enhance the beneficial effects of THC. Other cannabinoid and non-cannabinoid compounds in herbal cannabis or its extracts may reduce THC-induced anxiety, cholinergic deficits, and immunosuppression. Cannabis terpenoids and flavonoids may also increase cerebral blood flow, enhance cortical activity, kill respiratory pathogens, and provide anti-inflammatory activ-ity. [Article copies available for a fee from The Haworth Document Delivery Service: and: Cannabis Therapeutics in HIV/AIDS (ed: Ethan Russo) The Haworth Integrative Healing Press, an imprint of The Haworth Press, Inc., 2001, pp. 103-132. Single or multiple copies of this arti-cle are available for a fee from The Haworth Document Delivery Service [1-800-342-9678, 9:00 a.m. -5:00 p.m. (EST). E-mail address: getinfo@haworthpressinc.com].
Article
A protein determination method which involves the binding of Coomassie Brilliant Blue G-250 to protein is described. The binding of the dye to protein causes a shift in the absorption maximum of the dye from 465 to 595 nm, and it is the increase in absorption at 595 nm which is monitored. This assay is very reproducible and rapid with the dye binding process virtually complete in approximately 2 min with good color stability for 1 hr. There is little or no interference from cations such as sodium or potassium nor from carbohydrates such as sucrose. A small amount of color is developed in the presence of strongly alkaline buffering agents, but the assay may be run accurately by the use of proper buffer controls. The only components found to give excessive interfering color in the assay are relatively large amounts of detergents such as sodium dodecyl sulfate, Triton X-100, and commercial glassware detergents. Interference by small amounts of detergent may be eliminated by the use of proper controls.
Article
This chapter discusses measurement of receptor-stimulated guanosine 5'-O-(γ-Thio)triphosphate binding by G Proteins. Many transmembrane signaling processes caused by extracellular hormones and neurotransmitters are mediated by receptors interacting with heterotrimeric (αβγ) guanine nucleotide-binding proteins (G proteins) attached to the inner face of the plasma membrane. Agonist-liganded receptors apparently initiate activation of G proteins by catalyzing the exchange of guanosine 5'-diphosphate (GDP) by guanosine 5'-triphosphate (GTP) bound to the α subunits. In membrane preparations and reconstituted systems, this activation process is frequently monitored by studying agonist stimulation of high-affinity GTPase, an enzymatic activity of G-protein α subunits. To study the initial steps of G-protein activation by agonist-liganded receptors in a quantitative manner, the binding of radiolabeled GTP analogs, which are not hydrolyzed by the GTPase activity of G-protein α subunits, to G proteins is determined. Of these GTP analogs, guanosine 5'-O-(γ-[35S]thio)triphosphate ([35S]GTPγS) is most frequently used. This nucleotide has a high affinity for all types of G proteins and is available with a relatively high specific radioactivity (1000-1400 Ci/mmol; physical half-life 87.4 days). The chapter describe the measurement of receptor induced binding of [35S]GTPγS to membranous and detergent-solubilized G proteins and how this method can be adapted to different G proteins for an optimal response to receptor stimulation.
Article
(³H)Spiroxatrine was examined as a potential ligand for the labeling of 5-HT/sub 1A/ sites in the rat hippocampus. Analysis o the binding of (³H)spiroxatrine in the absence and presence of varying concentrations of three monoamine neurotransmitters revealed that serotonin (5-HT) had high affinity for the (³H)spiroxatrine binding sites, consistent with the labeling of 5-HT⁠sites, while dopamine and norepinephrine had very low affinity. Saturation studies of the binding of (³H)spiroxatrine revealed a single population of sites with a K/sub d/ = 2.21 nM. Further pharmacologic characterization with the 5-HT/sub 1A/ ligands 8-hydroxy-2-(di-ni-propylamino)tetralin, ipsapirone, and WB4101 and the butyrophenone compounds spiperone and haloperidol gave results that were consistent with (³H)spiroxatrine labeling 5-HT/sub 1A/ sites. This ligand produced stable, reproducible binding with a good ratio of specific to nonspecific binding. The binding of (³H)spiroxatrine was sensitive to GTP, suggesting that this ligand may act as an agonist. 21 references, 5 figures, 2 tables.
Article
Serotonin1A (5-HT1A) receptors are located on both 5-HT cell bodies where they act as inhibitory autoreceptors and at postsynaptic sites where they mediate the effects of 5-HT released from nerve terminals. The sensitivity of 5-HT1A receptors in humans can be measured using the technique of pharmacological challenge. For example, acute administration of a selective -HT1A receptor agonist, such as ipsapirone, decreases body temperature and increases plasma cortisol through activation of pre- and postsynaptic 5-HT1A receptors, respectively. Use of this technique has demonstrated that unmedicated patients with major depression have decreased sensitivity of both pre- and postsynaptic 5-HT1A receptors. Treatment with selective serotonin reuptake inhibitors further down-regulates -HT1A receptor activity. Due to the hypotheses linking decreased sensitivity of 5-HT1A autoreceptors with the onset of antidepressant activity, there is current interest in the therapeutic efficacy of combined treatment with selective serotonin reuptake inhibitors and -HT1A receptor antagonists.
Article
Serotonin1A (5-HT1A) receptors are located on both 5-HT cell bodies where they act as inhibitory autoreceptors and at postsynaptic sites where they mediate the effects of 5-HT released from nerve terminals. The sensitivity of 5-HT1A receptors in humans can be measured using the technique of pharmacological challenge. For example, acute administration of a selective 5-HT1A receptor agonist, such as ipsapirone, decreases body temperature and increases plasma cortisol through activation of pre- and postsynaptic 5-HT1A receptors, respectively. Use of this technique has demonstrated that unmedicated patients with major depression have decreased sensitivity of both pre- and postsynaptic 5-HT1A receptors. Treatment with selective serotonin reuptake inhibitors further down-regulates 5-HT1A receptor activity. Due to the hypotheses linking decreased sensitivity of 5-HT1A autoreceptors with the onset of antidepressant activity, there is current interest in the therapeutic efficacy of combined treatment with selective serotonin reuptake inhibitors and 5-HT1A receptor antagonists.
Article
The recent cloning of the complementary DNAs and/or genes for several receptors linked to guanine nucleotide regulatory proteins including the adrenergic receptors (alpha 1, alpha 2A, alpha 2B, beta 1, beta 2), several subtypes of the muscarinic cholinergic receptors, and the visual 'receptor' rhodopsin has revealed considerable similarity in the primary structure of these proteins. In addition, all of these proteins contain seven putative transmembrane alpha-helices. We have previously described a genomic clone, G-21, isolated by cross-hybridization at reduced stringency with a full length beta 2-adrenergic receptor probe. This clone contains an intronless gene which, because of its striking sequence resemblance to the adrenergic receptors, is presumed to encode a G-protein-coupled receptor. Previous attempts to identify this putative receptor by expression studies have failed. We now report that the protein product of the genomic clone, G21, transiently expressed in monkey kidney cells has all the typical ligand-binding characteristics of the 5-hydroxytryptamine (5-HT1A) receptor.
Article
Migraine is a frequent paroxysmal headache disorder of unknown aetiology. Genetic factors may control attack frequency and possibly attack severity. Serotonin1D (5-HT1Dβ) receptors have a prominent position within the final common pathway of the mechanisms involved in the headache and associated symptoms. Stimulation of these receptors by selective 5-HT1Dβ receptor agonists such as sumatriptan and newer compounds including MK-462 and 311C90, rapidly and fully blocks the symptoms of the headache phase. The efficacy depends on factors such as timing of administration during or before the headache, speed of initial rise of drug plasma levels, and possibly degree of brain penetration. All agonists at S-HT1Dβ receptors share a short duration of action resulting in recurrence of the headache symptoms within 24 h in about one-third of attacks in clinical trials. The risk for headache recurrence seems patient dependent: about 10% of patients treating multiple attacks experience headache recurrence in every treated attack, whereas 40% never experience recurrence. These differences are not related to simple pharmacokinetic differences between patients or drugs. Increasing plasma half-life of the drug will most likely not reduce the risk of recurrence. “Breakthrough of peripheral suppressive effect” with an ongoing “central migraine generator”, rather than the occurrence of a new attack, seems to be the most likely underlying mechanism for headache recurrence. In a minority of, possibly predisposed, patients, use of sumatriptan may induce increase of attack frequency. Four mechanisms have been suggested for the antimigraine action of 5-HT1Dβ receptor agonists: (1) vasoconstriction of cranial, most likely meningeal and dural blood vessels; (2) inhibition of release of vasoactive neuropeptides from perivascular trigeminal nerve terminals within dura mater and meninges; (3) blockade of trigeminal nerve terminal depolarization; and (4) central inhibition within the trigeminal nucleus caudatus in the brainstem. Which of these mechanisms is the most important, and whether or not vasoconstrictor action is necessary for antimigraine efficacy, is currently under extensive investigation. At this point all drugs with proven antimigraine efficacy share the ability to contract blood vessels and thus all feature also the potential risk of causing vasoconstriction of coronary vessels. In relation herewith, major efforts are put into the search for “the antimigraine receptor” and which receptor subtype mediates which action of sumatriptan-like drugs. At this point, the 5-HT1Dβ receptor subtype is thought to mediate vasoconstriction. Some investigators feel that the 5-HT1D receptor subtype mediates the neuronal effects of sumatriptan, while others are much less convinced about the physiological role of this subtype of receptor. Further research into receptor subtype specificity and affinity of compounds may promote the development of even better antimigraine drugs.